Shared Flashcard Set

Details

Pharmacology (Midterm)
UCLA nursing
188
Pharmacology
Graduate
02/07/2012

Additional Pharmacology Flashcards

 


 

Cards

Term
What is pharmacology?
Definition
It is the science of drug action on biology system... while Drug is any chemical that can affect living process.
Term
What are the properties of ideal drug?
Definition

1. Effectiveness

2. Safety

3. Selectivity

Term
Which  property of drug is the most imfortant one?
Definition
Effectiveness because it's no point to use the drug if it's not gonna do its job!
Term
What is a "Therapeutic objective"?
Definition
To provide maximum benefit with minimum harm
Term
Other properties of drug (beside those 3)?
Definition

1. Reversible action--> ex. Anesthetic med

2. Predictability

3. Ease of administration--> improve compliance (#dose/day)

4.Freedom for drug reaction

5. Low cost

6. Chemical stability--> drug should be stable!

7. Possession of simple generic name

Term
Factor influencing the intensity of drug response?
Definition
[image]
Term
Stages of new drug development?
Definition

1. Preclinical testing (1-5 yrs)

*** test in animals**

*** test for toxicity, pharmacokinetic properties, useful effects.**

2. Clinical testing (2-10 yrs)

 

Term

What group of pp will be tested on during phase I?

a. Healthy volunteer

b. Patients with diease

c. Large group of patients with disease

d. Large group of healthy people

Definition

Answer A!

Healthy people (25-50 pp)

Test for: metabolism & effects in human

Term

         Clinical testing : Phase 2?

Definition
  • Determine efficacy and dosage range
  • Test on 100-200 pp
Term
Clinical testing: Phase 3?
Definition
  • Larger population
  • Tested in 1000s patients with disease
  • Tested for establish safety and feectiveness
  • Submission of new drug application (NDA) to the FDA
Term
Clinical testing: Phase 4
Definition
  • Post-marketing suveilance
Term
What are the limitation of Clinical testing procedure?
Definition
  • Limited info. for women and children
  • Faliure to detect all adverse effects:

1. Relatively small number of pts.

2. Pts carefully selected in trials and do not represent full spectrum

3. Pts in trails take drug for relatively short period of time.   

Term

What are three types pf drug names?

Definition
  1. Chemical name
  2. Generic name *** Prefer!
  3. Trade name
Term

Which name to use--Generic or Trade?

Definition
  • Problem with generic names are
      * 4.3 syllables, trade name 2.3 
          syllabus
      * More difficult to remeber
  • Problem with trade names are:
      *A single drug can have multiple trade names
      * Same trade name can use for diferrent product.
Term
**Are generic products and brand-name products therapeutically equivalent?
Definition
  • Contain the same dose of the same drug
  • FDA approved generic as equal
  • Rate and extent of absorption may differ and may be a concern for drug wuth a narrow therapeutic window
Term

 

What is pharmacokinetics?

a. What the drug does to the body

b. What the body does to the drug

Definition

Answer B.

Study of the absorption, distirbution, metabolism, and excretion of drug ("ADME")

Term

 

ADME!

Definition
[image]
Term

 

Defines "Absorption"?

Definition
Absoption is the movement of a drug from its site of administration into the blood.
Term

 

Defines "Distribution"

Definition
Drug movement from the blood to the interstitial space of tissues and from there into cells
Term
Defines "Metabolism"
Definition
Biotransformation!---> enzymtically mediated alteration o drug structure.
Term

 

Defines "Excretion"

Definition
The movement of drugs and their metabolites out of the body.
Term
How do molecules cross cell membrane?
Definition
  1. Channel and pore--> very few drug can cross this membrane (weight<200 can cross through)
  2. Transport system
  3. Direct penetration of the membrane--> "Passive diffusion"
Term
What is the most common way for drugs to cross cell membrane?
Definition

Passive diffusion

  • Movement of substance from higher concentration to lower concentration.
  • Drug must be lipid soluble to cross a membrane
Term

 

pH-dependent ionization

Definition

Most drugs are weak acids and bases --->ionization is determined by the pH of biological fluid

Term

 

Ion trapping of drugs

Definition

- Occur when pH of biological fluids are different in each side of the cell membrane (i.e. stomache or plasma)

- The ionized form of a drug is trapped in the side where the pH most favors the drug ionization.

Term

Tell me more about 

" Ion trapping!"

Definition

Acidic drug--> ionize in basic media--> will accumulate in basic (alkaline) side

Basic drug--> ionize in acid media--> will accumulate in acidic side.

Term

What kind of drugs will be ion-trapped in the renal ultrafiltrate if the urine pH is lowered by large doses of ascorbic acid (Vit. C)?

a. Weak acid

b. Weak base

Definition
Answer B ;because the situation is analogous to stomach, so weak base would be ion-trapped in urine.
Term

             

               Rate of absorption?

Definition
To determine how soon effects will begin
Term

 

         Amount of absorption?

Definition
To determine how intense effects will be.
Term
Will a weak acid or a weak base have a higher concentration in the stomach?
Definition

Answer Weak basic!

Because weak basic ionize in acid media, and stomach is very acidic (~2)

Term
What are the factors that affect drug absorption?
Definition
  1. Rate of dissolution --> form and route of drug (i.e. syrup, capule)
  2. Surface area ---> "large surface, better absorption"
  3. Blood flow--> More rapidly absorbed from sites where blood flow is high
  4. Lipid solubility--> Highly lipid soluble drugs are absorbed more rapidly
  5. pH partitioning--> Enhanced absorption of drugs that will be preferentially ionized in the plasma.
Term
Q. Why adverse effects are often not detected during each of the 4 phases of drug during evaluation in humans?
Definition

Failure to detect all adverse effects:

1. Due to small amount of pts.

2. Pts. carefully selected in trails and do not represent full spectum.

3. Pts. in trails take drug for a reletively short period of time

Term
Q. Why is it so important for us to know about "Pharmacokinetic"?
Definition
Because it can help maximum benefits and minimize harm.
Term
Q. What determines the intensity of drug response? What lies between the dose and intensity of response?
Definition

1. Prescribed drug--> dose given, error, route, timing, adherence.

2. Administered dose--> Pharmacokinetic (ADME)

3. Concentration at site of action--> Pharmacodynamicthe drug's impact on the body (drug-receptor interaction, patient’s functional state, and placebo effects)

Term

Q. Which part of cell membrane is hydrophobic?

a. Head

b. Tails

Definition

Answer is "TAILS" --> Hydrophobic is water-hating  !  

 

[image]

Term
What characteristics of compound allow them to diffuse between body compartment?
Definition
Nonpolar, nonionized, lipid-soluble, small particle.
Term
Which mechanism is the most common one for drug to cross cell membrane?
Definition

Passive diffusion--> From higher concentration to lower concentration.

***However, drug must be lipid solution in prior to cross the membrane.***

Term
Q. How does the ionization of a drug affect its availibility for action?
Definition
The ionized drug cannot pass through the membrane but the nonionized drug can.
Term
Q. What are four basic pharmacokinetic?
Definition
[image]
Term

 

Concept of ion trapping

Definition

Weak acid will be trapped (ionized) in basic media which makes weak acid no longer able to cross or be absorbed! Remember, weak acid will get absorbed in acid area.

Weak base will be trapped (ionized) in acidic media which make weak basic no longer able to cross membrane. Remember, weak basic will get absorbed in basic environmet.

Term
Why the acid drug like ASA have to cross from stomach to plasma?
Definition
Becasue it follows the law of diffusion. It tries to reach the equilibrium on both side of the membrane. 
Term
Q. Will a weak acid or a weak base have a higher concentration in the stomach?
Definition
Weak base---> weak base will be ionized in acidic media. Therefore, basic concentration will be high in the stomach
Term
Q. Will a weak acid or a weak base be eliminated more effectively in the alkaline urine.
Definition
Weak acid
Term

Matching

__1. Rate of absorbtion  

__2. Amount of absorption

a. how intense effects will be 

b. how soon effects will begin

Definition

Answer...

1. B

2. A

Term
What are the factors that affecting drug absorption?
Definition

1. Rate of dissolution--> Dissolve fast, onset fast (syrup, capsule)

2. Surface area--> Larger surface=faster absorption (i.e. small intestine has a better absorption bcoz of its microvilli)

3. Blood flow--> More rapidly absorbed from sites where blood flow is high

4. Lipid solubility--> Highly lipid soluble drugs are absorbed more rapidly.

5. pH partitioning--> 

Term
Why the route of administering drugs is important?
Definition
Because route affects onset and intensity of drug effects
Term
What is a first-pass elimination?
Definition

It is a following absorption portal blood delivers drug to liver.

**Drugs the are metabolized efficiently by the liver---> removed from circulation before distribution to the   body.  Also, rapid hepatic inactivation of certain oral drugs due to capacity of liver to metabolize certain oral drugs, inactivating drugs on first pass thru liver = drug has no therapeutic effect

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Term
Why does the dose of oral drug have to be larger than parenteral drug?
Definition
Because some drugs may get metabolized in the liver before reaching its site of action, so not all of the adminited dose reach the site of action.
Term
What is an Enterohepatic recirculation (recycling)?
Definition

1. Some oral drugs are excerted in to the bile and re-enter the small intestine via the bile duct.

** These drugs may:

1. Be absorbed into the portal blood and create a cycle of enterohepatic recirculation

2. Exit the body in the stool

 

Term

 

Tablets !

Definition

**Chemical equivalence: 2 preparations that contain the same amount of the identical compound.

**Bioequivalence: drug is absorbed at the same rate & same extent

***Note: formulation may be chemically equivalent but not bioequivalent.

Term
Where most drug metabolism takes place?
Definition
Liver--> Major Drug metabolism
Term
What is the major drug excretion?
Definition
Kidney---> Getting rid of the drugs from the body preventing drug accumulative, which can cause toxicity.
Term

Where is the site of absorption of enteric coated drugs?

a. Intestine

b. Stomach

3. Both

Definition
Answer is A. ---> TO protect drug and also stomach
Term
Q. How would a first-pass effect alter the bioavailiability of a drug after IV administation?
Definition
  • To circumvent first-pass effect, drug that undergoes rapid hepatic metabolism often administered parenterally (e.g. IV)
  • This allows drug to temporarily bypass the liver, allowing it to reach therapeutic levels in systemic blood
Term
Q. How woulda first-pass effect alter the bioavailibility of drug after oral administration?
Definition
  • Sublingual administration effective because permits nitroglycerin to be absorbed directly into systemic circulation
  • In circulation, drug carried to sites of action prior to passage thru liver, so that therapeautic action can be exerted before drug is exposed to hepatic enzymes
Term
Why will a slowly eliminated active metabolite make a significant contributions to the overall effect of initial drug after multlple dose?
Definition

Because "enterohepatic recirculation" delays elimination of the initial drug dose at the same time as later doses are being administered → prolongation of drug effect

Term
Which route is te best route for emergencies requiring rapid onset (code blue drug)?
Definition

IV (parenteral)

Term

Pharmacokinetic

"Blood flow"

Definition
  • In the first phase of distribution,  drugs are carried by the blood--> target (tissue, organ)
  • Blood flow determines rate of distribution (& rate of absorption--recall)
Term

 

Defines "Distribution"

Definition
The movement of drugs throughout the body 
Term
What are three major factors of drug distribution?
Definition

1. Blood flow to tissue

2. The ability of drug to exit the vascular system

3. The ability of drug to enter the cell.

Term
Q. Why can't the antibiotic drug treat the undrained absesses?
Definition
Because inside the undrained absesses don't have a vascular, which is important to carry the drug. Antibiotic will work if the absesses are drained.
Term

 

Exiting the Vascular system

Definition

1. Typical capillary beds

2. BBB

3. Placental drug transfer

4. Protein binding

Term
Why does capillary bed offer no resistance to the exiting of the drug from vascular system?
Definition
Because the drugs can sinply pass through the pore of capillary well not through the cell
Term
What kind of drug can pass through BBB from vascular system?
Definition

Only lipid soluble drug! 

BBB- Tight junctions between cells that compose the walls of capillaries in the CNS prevent drugs from passing between cells to exit the vascular system. Therefore, drug must pass directly through cells of the capillary wall, which means drug must be lipid soluble in order to pass through cell.

Term

 

Placental drug transfer

Definition

Placenta membrane seperate the fetal circulation to the maternal circulation--> do not constitute an absolute barrier to the passage of drugs--> same rules as for other membranes.

 

[image]

Term
Would a decrease in protein binding (caused by displacement by another drugs or reduced serum albumin) be expected increase or decrease the effect of a given dose of a highly bound drug?
Definition

Answer is to increase

  • Plasma albumin is one of the most important proteins a drug can bind to.
  • Due to this protein’s large size, it is unable to exit the bloodstream.
  • If there is a strong attraction (affinity) between albumin and the drug, the drug will bind to albumin and will be unable to leave the bloodstream, not be able to reach its site of action, metabolism or excretion.
  • Also, albumin has a limited number of binding sites and drugs compete with one another for binding sites that may result in displacement of another drug.
  • As a result, the displaced drug will be able to exit the bloodstream because it is no longer bound to the plasma albumin and enter the tissues where the concentration within the tissue increase.
  • Thus, the competition for binding can increase the intensity of drug responses ~> side effects and toxicity can result.
Term

Q. Which one of these two drug will more likely to reach the target tissue?

a. Bound drug (bound with protein binding, albumin)


b. Unbound drug

Definition

Answer is A.

 

Term

 

Defines Metabolism

Definition
Known as biotransformation, is defined as the enzymatic alternation of drug structure. Mostly takes place in LIVER
Term
What are six possible consequences of drug metabolism?
Definition

1. Accelerated renal drug excretion

2. Drung inactivition

3. Increased therapeutic action

4. Increased toxicity

5. Decreased toxicity

Term

 

CYP enzyme famillies

Definition
CYP 1, 2, 3 enzyme families
CYP 4-12 metabolize endogenous compounds
Asians are poor metabolizers of CYP2D6
Ethopians have ultra fast metabolisms
CYP3A4 is most abundant, 60% of metabolism
Term
Wht is  "PHARMACOGENETIC"?
Definition
CYS enzymes are genetically regulated---> genetic difference in metabolism
Term

Which CYS enzyme is the most abundant, reponsible for 60% of metabolism?

a. CYP2D6

b. CYP3A4

c. CYP3D3

Definition
Answer is  B
Term

Matching

__1. Phase 1

__2. Phase 2

a. Functionalization

b. Conjugation

Definition

Answer 1. A

             2. B

Term
How does the drug metabolism accelerate renal drug excretion?
Definition
Since kidney (the major of excretion) can't get rid of lipid soluble drug, at the liver the enzyme converts the lipid soluble to polar molecule (oxidation)
Term
What is an Enzymatic Inducers?
Definition
  • Increases the metabolism of drugs
  • Decrease drug absorption--> think of women taking oral contraceptive and phenobarbital at the same time. 
  • The inducer makes the drug loss their effectiveness.
  • Decrease drug plasma.
Term
What is an Enzymatic Inhibitors?
Definition
  • Inhibit the metabolism if drug
  • Increase drug plasma
  • Drug is more likely effective.
Term
When metabolic enzymes are induced, would you expect the dose of affected drugs to need to be increased or decreased?
Definition

Answer Increase.

Note:

Inducers: increase the metabolism of drugs causing…

  • Decrease drug concentration in plasma
  • Decrease half lives of drugs
  • …thus, the dose of the affected drugs need to be increased.
Term

When metabolic enzymes are inhibited, would you expect the dose of affected drugs to need to be increased or decreased?

Definition

Answer Decrease.

Note:

 

Inhibitors: inhibit the metabolism of drugs causing…

  • Increase drug concentration in plasma
  • Increase half lives of drugs
  • …thus, the dose of the affected drugs need to be decreased.

 

Term

 

Defines Excretion

Definition
  • Removal of drugs from the body
  • Urine pH
  • Ionized metabolites are excreted
  • Age, pathology--> change excretion
  • Drugs and metabolites can exit the body in urine, bile, sweat, saliva, breast milk, and expire air
Term

Which one is a renal route of drug excretion?

a. Bile

b. Sweat

c. Urine

d. Breast milk

Definition
Answer C
Term

Q. Which one is not correct about Excretion?

a. Nonionized metabolites are excreted

b. Age can pathology can change excretion

c. Nonrenal routes of drug excretion are saliva, breast milk, sweat.

Definition

Answer A.

Ionized drugs are excreted.

Term
What are three steps in renal drug excretion?
Definition

1. Glomerular filtration

2. Passive tubular reabsoption

3. Active tubular secretion

[image]

Term
What is Glomerular Flitration?
Definition
  • Drugs bound to albumin are not filtered
  • Most drug cross freely
Term

What is Passive tubular reabsorption?

 

Definition
  • Bcoz concentration in blood is lower than in the tuble
  • Lipid soluble drugs move along this gradient back into blood
  • Ions and polar drugs are excerted in urine.
  • By converting lipid soluble drug to more polar form, drug metabolism reduce passive reabsorptionof drugs and thereby accelerates their excretion. 
Term
What is Active tubular secretion?
Definition
Active transport system in the tubles that pump drugs from the blood to the tubular urine. Acids and bases are pumped, and P-glycoprotein also pumps drugs into the urine.
Term
What kinds of drugs are passively reabsorbed?
Definition
Lipid solution---> reabsorbed back to the blood from tubles
Term
What kinds of drugs are actively secreted (Renal machanism)?
Definition
Acid and Base
Term
What kind of drugs are rapidly excreted?
Definition
Ion and polar drugs
Term
Time course of drug response
Definition
  • To achieve the therapeutic objective, we must control the time course of drug response
  • Time course of drug response is directly related the concentration of drug in the blood
Term
What is a plasma drug level?
Definition
  • 2 types--> minimum effective concentration (MEC), toxic concentration
  • Clinical significance of plasma drug levels
Term
What is a Therapeutic range?
Definition
  • Range between the MEC and the toxic concentration
  • To maintain plasma drug levels within the therapeutic range.
Term

In the single-dose time course, the duration of effects is determined largely by the combination of.........?

 

Definition

Answer Metabolism and Excretion

[image]

Term

Which one tends to have a 100% bioavailibility?

a. IV dose

b. Oral dose

Definition

Answer A. 

Bioavailibility is proportion of administered dose present in plasma

Because IV dose is assumed to have a bioavailibility of 100% while oral dose usually <100% (incomplete)

 

Term
What are three pharmacokinetic parameters?
Definition

1. Apparent volume of distribution

2. Clearance

3. Half life (t1/2)

Term
What is a VD?
Definition
  • Volume of distribution
  • VD= total amount of drug in the body/ [drug plasma]
  • Can exceed any physical volume bcoz it is the voloume apparently necessary to contain the amount of drug homogenerously at the concentration found in plasma.
Term
Drugs with high VD have higher concentrations in............ than in the............?
Definition
Answer extravascular tissue than blood plasma.
Term
Is the drug with a high volume of distribution more lipid-soluble or more water-soluble?
Definition
Answer more lipid soluble bcoz it can cause the blood vassel to tissue.
Term
Would an antibotic with low volume of distribution be a good choice for an infectionon the toe of a diabetic pt?
Definition
Answer No, it's not a good choice because the drug with low Vd cannot cross to the tissue.
Term

Which would you expect to be cleared more rapidly?

a. drug with high Vd

b. drug with low Vd

Definition

Answer B. 

Becasue the drug with low Vd is likely polar and lipidphobic.

Term

 

Defines "Clearance (CL)"

Definition
  • measure of the ability of the body to eliminate drug
  • CL = Rate of elimination/ [C]

*** C stands for plasma drug concentration

  • First order elimination
  • For most drug, rate of eliminatin is directly propertionl to drug concentration
Term
The equation for clearance shows that it is determined by what two things?
Definition
Rate of elimination and plasma drug concentration [C]
Term
Clearance is affected by blood flow to which organ?
Definition
Heart, kidney, liver
Term
What is a capacity-limited elimination Saturation)?
Definition
    • For a few drugs, elimination becomes saturated at high concentration and is independent of concentration.
      • Ethanol, phenytoin, Aspirin
    • In this case, the rate of elimination and clearance of drug is no longer related to concentration.
      • Steady state not achieved; plasma concentration can increase unpredictability if dose is increased.
      • Clinically important when changing dosage of drugs with narrow therapeutic window.
Term

 

Drug Half-Life

Definition
  • t1/2: time for drug level to decline by 50%
  • Depends on both clearance and volume of distribution
Term
After how many half-lives , drund will turn to steady state of independent of dose?
Definition
Answer 4-5 half-lives
Term
Which clinical issue might be associated with drugs with long half-lives?
Definition
If a drug is toxic and has a longer half-life, then it will spend a longer time in the body system and have greater toxic effects.
Term

 

Short half-lives?

Definition

 

    1. If short half-life, the dosing interval must also be short. If a long dosing interval were used, drug levels would fall below the minimum effective concentration between doses, and you’d lose the therapeutic effect of the drug.
      1. If the patient is not compliant and doesn’t take the doses as scheduled and the drug has a short half-life, then the drug will not be at high enough concentrations to work.
      2. That’s why some drugs can’t just be taken once a day, need multiple doses/day.

                          b.  If long half-life, a long time can separate doses without loss of effect.

 

Term
What is a capacity-limited elimination?
Definition
  • For a few drugs, elimination becomes saturated at high concentration and is independent of concentration (ie. aspirin)---> rate of elimination and clearance is no longer related to concentration.
  • Steady state not achieved; plasma concentration can increase unpredictably if dose is increased.
Term

 

Loading dose

Definition
For drugs with half lives it may take too long (days or weeks) to reach steady state ---> Administration of loading dose to reach target concentration prompty
Term
What is the effect of a loading dose on the speed with which steady state is reached?
Definition
Loading dose helps the long half lives drugs to reach the steady state faster. However, these long half lives drugs still need to take smaller dose to maintain the stead state.
Term
What effect would the inhibition of metabolism have on the action of a drug whose elimination is metabolism-dependent?
Definition
  • On the action of a drug whose elimination is metabolism-dependent -- a metabolism-dependent drug is ingested in a “prodrug” state and only becomes active post-metabolism; therefore metabolism inhibition would reduce the drug’s action
Term

 

Dose-response relationship

Definition
  • To understand drug-receptor interaction, we need to quantify the relationship between the drug and the biological effect it product
  • "Dose-response curve"
Term

 

Maximum efficacy?

Definition
  • Maximum efficacy= largest effect a drug can produce
  • It is indicated by the height of the dose-response curve
**** Remember, it's the most important of an ideal drug
Term

 

Potency

Definition

Potency is how much drug must be administered to elicit the response

[image]  In this case taking a pain relief with meperidine requires higher doses than morphine.. THerefore, morphine is more potent than meperidine.

Term

 

Pharmacodynamics

Definition
  • Based on receptor theory: a drug must first be bound to receptive substance in order to produce an effect.
  • Drug targets --> describes the cellular molecule to which a drug binds to initiate its effect.
Term
What is an EC50?
Definition
Potency refers to the concentration (EC50) or dose (ED50) of a drug required to produce 50% of that drug's maximal effect.
Term
If drug A has a higher ED50 than drug B, is it more or less potent?
Definition
Answer less potent because drug A requires a higher dose to produce the defined therapeutic response compared to drug B
Term
What are 4 primary receptors?
Definition
Cholinergic, adrenergic, dopaminergic, serotoninergic.
Term

 

Affinity ?

Definition

Affinity : Strength of the attraction between the drugs and its receptor---> tendency of drug to bind to receptors.

** Drug with high affinity are very potent

Term

 

Intrinsic activity

Definition
  • The ability of a drug to activate its receptor upon binding
  • Drug with  intrinsic activity cause intense receptor activation high
  • The intrinsic activity of a drug ---> reflected in its efficacy
Term
What is a drug-receptor binding?
Definition
Chemical bonds that form the binding interaction between drug and target.
Term

Drug-receptor binding

" Type of binding "

Definition

Ionic--> interact between charge, strong bond

Hydrogen--> b/t H atoms & pairs of free electron on O and N atoms

Forces are summed: more bonds = better fit and higher affinity.

Term
After the drug is bonded with receptor, is it impossible for the reaction to be reversible?
Definition

Yes! binding interaction is usually reversible

However, it can only be irreversible if electrons are share.

Ex. ASA binds covanlently to COX--> causing plateles inhibited for the life of it.

Term
Does partial agonist have a high intrinsic activity?
Definition

No

Partial agonist --> acts on same receptor but no maximun effect

  • Has affinity but no intrinsic activity
  • Many drugs used clinically as antagonists are actually partial agonists--> mixed effects (agonist and antagonist)
Term

Which one in the following has affinity but not intrinsic activity?

a. Antagonist

b. Partial agonist

c. Full agonist

d. a,b are correct

Definition

Answer is A.

However, partial agonist has less intrinsic activity than full agonist

Term
Why will you never suggest your pt to take antibiotic (ie. penicilins) with juice?
Definition
Because acidity from fruit juices may decompose penicilins, erythromycin.
Term
What inhibits CYP3A metabolism?
Definition
Grapefruit juice inhibits CYP3A metabolism---> single 8oz glass inhibits for24-48 hrs lol
Term

Pharmacokinetic

"Neonates & infants"

Definition
  • Absorbtion: prolonged and irregular gastric emptying (pH>4)
  • Distribution: protein binding limited--> increased free drug & intensified effects
  • BBB not fully developed
  • Hepatic metabolism: metabolizing capacity low
  • Renal excretion: Renal blood flow & glomerular filtration low
Term
Pharmacokinetics chnages in the elderly
Definition
  • Absorption--> rate of absorption may be slow
  • Distribution--> increase %body fat, decrease %lean body mass, decrease total water, reduce concentration of serum albumin 
  • Metabolism (increase t1/2)
  • Excretion---> drug accumulation secondary to reduced renal function.
Term

 

Antiviral Therapy

Definition
  • Ability to treat viral infection remains limited
  • Viruses use biochemical nachinery of host cells to reproduce --> difficult to suppress viral replication w/o doing harm to the host
  • Anitvirals suppress biochemical processes unique to viral reproduction.
Term
Whst is th drug for HSV and VZV?
Definition

Prototype: Acyclovir [Zovirax]

Similar drug: Valacyclovir (prodrug of acyclovir)

** This drug family ends with -cyclovir

Term

 

MOA of HSV and VZV

Definition
  • Acyclovir inhibits viral replication by suppressing synthesis of viral DNA
  • Inhibition of viral DNA polymerase 
  • Becoming incorporation into the growing viral DNA strand, which block further stand growth.
  • Acyclovir must be activiated by viral thymidine kinase then active species inhibit DNA synthesis.
Term

Clinical use of Acyclovir

for HSV and VZV

Definition

HSV-2 ---> po acyclovir decrease severity of initial episode, decrease frequency of recurrence... Does not eliminate the virus!

HSV-1 --> po acyclovir can be taken to prevent herpes labialis (cold sore)

VZV---> High doses are effective to treat shingle (in older adult), effective for varicella (in children)

** Oral acyclovir is deviod of serious adverse effects.

Term
What is a drug for treating CMV?
Definition
Ganciclovir--> only use to treat and prevent CMV
Term

What is a CMV?

Definition

CMV---->

  • Cytomegalovirus; member of herpes family
  • %0-85% of American age > 40 harbor the virus
  • Virus remains for life
  • Can be reactivated in immunosuppressive pt.
  • Same MOA as acyclovir
Term
What is Ganciclovir for?
Definition
For prevention and treatment of CMV
Term
Which hepatitis causes chronic hepatitis?
Definition

B, C, D

: Hepatitis C --> Transmission through blood

Term
What is the MOA of Interferon alpha preparation?
Definition
Binds to receptors on host cell membrane, blocks viral entry into the cell, synthesis of viral mRNA, and viral protein, and viral assembly and release
Term
What drug is used to treat Hepatitis C?
Definition
Answer Interferon alpha preparation
Term
What is the advantage of using peginterferon compared to conventional interferon?
Definition
Answer Peginterferon helps to delay elimination, lonf half-lives (1x/week)
Term
What is the efficacy of H1N1 vaccine?
Definition
Protection begins 1 to 2 weeks after vaccination nad last about 6 months.
Term
What are three classes of drugs active against influenza?
Definition

1. Influenza vaccine: Inactivated vaccine, Live Attenuated Influenza Vaccine (LAIV)

2. Neuraminidase Inhibitors: Oseltamivir [Tamiflu], Zanamivir [Relenza]

3. Adamantanes: Amantadine [Summetrel], Rimantidine [Flumadine]

Term

Which drug can treat both Inflenza A and Parkinson's disease?

a. Rimantidine

b. Amantadine

c. Acyclovir

d. Oseltamivir

Definition
Answer B. It's also called Symmetrel
Term

Which drug is a HIV fusion inhibitor drug?

a. Enfuvirtide

b. Ritonavir

c. Raltegravir

d. Zidovudine

Definition

Answer A. 

Enfuvirtide prevents HIV fusion with lymphocyte cell membrane--> block HIV entry and replication

Term
What is a HAART?
Definition

HAART is Highly Active Anti-Retroviral Therapy

3 or 4 drugs regimen:

1. Decrease plasma HIV to undetectable levels and delay loss of immune function

2. Expensive

3. Complex

4. Drug interaction

5. Must be lifelong

Term
Drug class selection used as HAART and the merits of combination therapy?
Definition

Preferred regimen have 3 drugs from different classes in order to:

1. reduce resistance

2. spare other classes for the future use.

** NNRTI-based regimen (1 NNRTI+ 2 NRTIs)

** PI-based regimen (PI+ NRTIs)

**INSTI-based regimen (INSTI+ 2 NRTIS)

Best way to memorize is that always (1)name of the base + (2) NRTIs

Term
What is the role of "basal ganglia"?
Definition

Inhibitory aspects of motor function

**Dysfunction causes tremor, rigidty, abnormal movement.

Term

 

How Drugs Affect the brain?

Definition

1. Drugs affect communication between neurons at the synapse (communication point)

2. Most drugs effect are based on specificity for neurotransmitter receptors in the brain--> overestimated, some drugs are not receptor specific

3. Neuronal activation results in excitatory/inhibitory signaling

     3.1 Inhibit of inhibitory receptor resulting in release from inhibition

4. Adapt receptor expression to long-term treatment

Term
What is Parkinson's diease?
Definition
Caused by degeneration of dopaminergic neuron in the substantia nigra  
Term
What are the motor symptom of Parkinson" disease?
Definition
  • Bradykinesia
  • Rigidity
  • resting tremor
  • postural instability, shuffling gait
*** Non-motor symptoms: conginative impairment, depression, constipation, urinary frequency/urgency, orthostatic hypotension.
Term
Which neuron in the substantis nigra become degenerated due to Parkinson's disease?
Definition
Dopamininergic neurons
Term
[image]
Definition

Dopamine Pathways

1. Nigrostriatal pathway--> Substantia Nigra to Striatum

: Motor control

: Death of neurons in this pathway can result in Parkinson's disease.

2. Mesolimpic &Mesocortical pathway---> Ventral Tegmental Area to Nucleus Accumbens, Amygdala &hippocampus, Prefrontal cortex

: Memory

: Motivation and emotional response

: Reward and desire

: Addiction

: Can cause hallucinations and schizophrenia

3. Tuberroinfundibular pathway---> hypothelamus to pituitary gland

: Hormonal regulation

: Maternal behavior

: Pregnancy

: Sensory process



 

Term

 

How can L-dopa get transported across the BBB?

Definition

L-dopa---> transported across BBB via amino acid transporter.

** Note!

  • immediate precursor of dopamine
  • converted to dopamine in brain by decarboxylase
  • 75% of pts get 50% reduction in symptom severity
  • Take several motns to get full effects, last for about 3-5 yrs
  • Best absorbed on empty stomach.
Term
Describe the possible sites of drug action on neurotrasmission
Definition

1. Modulate neurotransmitter release

2.Modulate neurotransmitter clearance from the synaptic cleft

3.Modulate effects of the neurotransmitter at the post-synaptic receptor

Term
Describe the mechanism of action for levodopa and carbidopa.
Definition
  • Levodopa mechanism of action
    • levodopa crosses the BBB via active transport (dopamine can not do this)
    • dopaminergic neurons in the striatum uptake the levodopa
    • levodopa is converted to dopamine via a decarboxylase, which is enhanced by pyridoxine (aka vitamin B6)
  • Carbidopa mechanism of action
    • without carbidopa, most of levodopa is converted to dopamine before it reaches the brain (98% of it)
    • carbidopa inhibits the carboxylase responsible for the conversion of levodopa to dopamine, but because carbidopa can not cross the blood-brain barrier, it selectively inhibits the carboxylases in the periphery (extra good because dopamine in the periphery causes cardiovascular responses + nausea/vomit), which brings down the portion of levodopa lost in the periphery from 98% to 90%
Term
Which class of drug may be useful in mild or early disease (Pakinson's disease)?
Definition

MAO- B inhibitor

  • Inhibit dopamine metabolism, prolong L-dopa effects
  • May have neuroprotective effects, so should be asministered  in the disease course early
Anticholinergic drugs
  • Restore baland between choligernic and dopaminergic
  • More useful in mild or early disease ** most effective for tremor & rigidity
Term

 

MOA of Entacapone

(Comtan)

Definition

** treatment for PD

MOA

1. Inhibits COMT (catechol-O-methyltransferase)

2. Increase plasma t1/2 of L-dopa, thus increase availibility

3. Available in combination with carbidopa/L-dopa

Term

Which dopaminergic drug helps to increase t1/2 of L-dopa?

a. Carbidopa

b. Entacapone

c. MAO-B

 

Definition

Answer B.

 

Term

 

MOA of dopamine aganist (D2)?

Definition
  • Use as monotherapy in youg pt: low risk of dyskinesias, may have neuroprotective effects
  • Adjunctive therapy with L-dopa
  • Ergot derivative--> older, less specific, more AE
  • Non-ergot derivatives (ie. apomorphine; administered SubQ)---> newer, more specific, better side effects
Term

 

MOA of MAO-B inhibitor

Definition

**Treatment for PD

*** selegiline, rasagiline

MOA

1. Inhibits dopamine metabolism, prolong L-dopa effects

2. Neuroprotective effects

Term

 

MOA of Amantadine

Definition

**Treatment for PD

1. Stimulate dopamine release, blocks dopamine re-uptake, blocks cholinergic and glutaminergic receptors

2. Most useful for dyskinesias

3. Short-lives

4. Significant AE --> insomnia, agitation, hallucination, restlessness

Term

What drug may provide some preventative effects if used early?

a. MAO-B inhibitor

b. L-Dopa

c. Amantadine

c. anticholinergic drug

Definition

Answer A.

 

Don't get confused with anticholinergic drug, which is more useful in mild or early disease.

Term

 

AE of L-Dopa (early)

Definition

Early

  • Early effects: nausea & vomiting in 80% of patients, but resolves with continued therapy
    • common anti-emetics relatively contraindicated because they have anti-dopaminertic function

 
 
 
 
 
 
Term

 

AE of L-dopa (Late stage)

Definition
    • wearing-off effect: same dose is less effective
    • on-off effect: rapid fluctuation between no effect and increased mobility but with disabling dyskinesias
    • with extended use, the therapeutic window for levodopa narrows, and effectiveness drops (the following effects can be seen early as well, but they’re more common later on as a result of this narrow therapeutic window)
      • dyskinesias, lots of involuntary movements
        • treat with reduction in L-dopa dosing
      • psychosis, visual hallucinations, paranoia
        • treat with clozapine (Clozaril), quetiapine (Seroquel)
      • postural hypotension
        • treat with fludrocortisone (Florinef) and/or midodrine (Proamatine)
Term
What are some strategies for dealing with loss of effectiveness problems after 3-5 years of therapy (in PD)?
Definition

 

  • begin treatment with dopamine agonists and save levodopa for later
  • initial strategy is to increase total dosing or dose frequency
  • use sustained release formulations to give stable level of levodopa for a long time
  • supplement with other PD medications
  • have the patient take a drug holiday to re-establish sensitivity
Term
Which drug is most effective in treating tremor and rigidity (in PD)?
Definition
Answer anticholinergic drugs
Term
Name another clinical use for the antimuscarinic trihexyphenidyl and benztropic
Definition
Movement or muscle tension disorders caused by antipsychotic medications
Term

 

Given Dopamine back

Definition

 L-dopa

 

Term

 Maximizing L-dopa delivery to the  brain?

Definition
Carbidopa, Entacapone
Term
Inhibits COMT and increase t1/2 of L-dopa?
Definition
Entacapone
Term
Restore balance between chlonergic and dopaminergic singnaling in basal ganglia?
Definition
Anticholinergic drugs
Term
Stimulate dopamine release, bolcks dopamine re-uptake, blocks cholinergic, glutaminergic receptors?
Definition
Amantadine (Suymmetryl)
Term

 

What causes Alzheimer's disease?

(AD)

Definition

** Amount of Ach is decreased; causing Alzheimer's 

** most common cause of dementia

1. Manifest clinically as progressive memmory loss

2. Initiated by amyloid acculation

    - Senile plaque and neurofibrillary tangles

    - Inflammatory response

 

    - Oxidative injuries & free radical formation

    - Neuronal loss in cortical and subcortical regions

Term
**What two drugs are most useful for control of problem behavior (in AD patient)?
Definition
Answer Risperidone (Risperdal) and Olanzapine (Zyprexa)
Term
Prevention approach that may slow decline in AD patients
Definition

1. Dietary supplements: Vit. E, gingko (not consistently effective)

2. Prevention: epidemiology supports NSAIDs, statin, curcumin, hormone replacement, fish oil

3. Drug in development: focused on inhibition of beta-amyloid synthesis.

Term
What is the name and MOA of the newest drug approved for treatment of AD?
Definition

**Memantine (Namenda)**

MOA

  • Blocks N-methyl-D-aspartate (NMDA) receptor ion channels
  • Inhibits calcium influx into neurons caused by excessive glutamate signaling --> reduce noise in learnign &memeory processes, prevent neurotoxiciy associated with excessive calcium influx
  • Better toleranted drug than AChe-Is
  • Most common AE --> lightheadache, confusion, headache
Term

 

MOA of "Tracrine and donepezil"

Definition

Tacrine & donepezil are centrally acting reversible cholinesterase inhibitors, leading to elevated Ach levels in the brain. The action slows the neoronal degradation that occurs in Alzheimer's disease

 

 

 

 

 

 

Term
What toxic compounds are related to tacrine and donepezil?
Definition
Donepezil & tacrine increase the effects of & risk of toxicity with theophylline and cholinesterase inhibitor. They decrease the effects of anticholonergic and increase the risk of gastrointestinal bleeding with NSAIDs.
Term

Name two limitaitons to these drugs as AD therapy

Definition
  • Tacrine has a high risk of liver injury and must be taken 4x/day (not a good idea for someone with AD).
  • Donepezil is more selective than tacrine, but can cause nausea and diarrhea, as well

    as bradycardia in patients predisposing to heart disease. Not hepotoxic as is tacrine.

Supporting users have an ad free experience!